Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
STAR Protoc ; 5(3): 103217, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39068648

RESUMEN

Study of disease-relevant immune cells, namely monocytes and macrophages, is limited based on availability of primary tissue, a limitation that can be remedied using human induced pluripotent stem cell (hiPSC) technology. Here, we present a protocol for differentiation of monocytes and macrophages from hiPSCs. We describe steps for hiPSC maintenance, mesoderm lineage induction, hematopoietic progenitor cells (HPCs) commitment and expansion, and myeloid lineage induction. We then detail procedures for monocyte formation and functional macrophage formation and polarization. For complete details on the use and execution of this protocol, please refer to Chen et al.1.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Macrófagos , Monocitos , Humanos , Células Madre Pluripotentes Inducidas/citología , Monocitos/citología , Macrófagos/citología , Diferenciación Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Células Madre Hematopoyéticas/citología , Células Cultivadas
2.
Front Immunol ; 15: 1288045, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38629065

RESUMEN

Thymic epithelial tumors (TETs) are rare mediastinal cancers originating from the thymus, classified in two main histotypes: thymoma and thymic carcinoma (TC). TETs affect a primary lymphoid organ playing a critical role in keeping T-cell homeostasis and ensuring an adequate immunological tolerance against "self". In particular, thymomas and not TC are frequently associated with autoimmune diseases (ADs), with Myasthenia Gravis being the most common AD present in 30% of patients with thymoma. This comorbidity, in addition to negatively affecting the quality and duration of patients' life, reduces the spectrum of the available therapeutic options. Indeed, the presence of autoimmunity represents an exclusion criteria for the administration of the newest immunotherapeutic treatments with checkpoint inhibitors. The pathophysiological correlation between TETs and autoimmunity remains a mystery. Several studies have demonstrated the presence of a residual and active thymopoiesis in adult patients affected by thymomas, especially in mixed and lymphocytic-rich thymomas, currently known as type AB and B thymomas. The aim of this review is to provide the state of art in regard to the histological features of the different TET histotype, to the role of the different immune cells infiltrating tumor microenvironments and their impact in the break of central immunologic thymic tolerance in thymomas. We discuss here both cellular and molecular immunologic mechanisms inducing the onset of autoimmunity in TETs, limiting the portfolio of therapeutic strategies against TETs and greatly impacting the prognosis of associated autoimmune diseases.


Asunto(s)
Miastenia Gravis , Neoplasias Glandulares y Epiteliales , Timoma , Neoplasias del Timo , Adulto , Humanos , Autoinmunidad , Neoplasias del Timo/complicaciones , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Glandulares y Epiteliales/complicaciones , Microambiente Tumoral
3.
Front Immunol ; 13: 1074762, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36703985

RESUMEN

Introduction: Adult-type diffuse gliomas are malignant primary brain tumors characterized by very poor prognosis. Dendritic cells (DCs) are key in priming antitumor effector functions in cancer, but their role in gliomas remains poorly understood. Methods: In this study, we characterized tumor-infiltrating DCs (TIDCs) in adult patients with newly diagnosed diffuse gliomas by using multi-parametric flow cytometry and single-cell RNA sequencing. Results: We demonstrated that different subsets of DCs are present in the glioma microenvironment, whereas they are absent in cancer-free brain parenchyma. The largest cluster of TIDCs was characterized by a transcriptomic profile suggestive of severe functional impairment. Patients undergoing perioperative corticosteroid treatment showed a significant reduction of conventional DC1s, the DC subset with key functions in antitumor immunity. They also showed phenotypic and transcriptional evidence of a more severe functional impairment of TIDCs. Discussion: Overall, the results of this study indicate that functionally impaired DCs are recruited in the glioma microenvironment. They are severely affected by dexamethasone administration, suggesting that the detrimental effects of corticosteroids on DCs may represent one of the mechanisms contributing to the already reported negative prognostic impact of steroids on glioma patient survival.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Adulto , Pronóstico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Corticoesteroides/uso terapéutico , Células Dendríticas , Microambiente Tumoral
4.
Neurooncol Adv ; 3(1): vdab160, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34901858

RESUMEN

BACKGROUND: Immunotherapeutic early-phase clinical trials (ieCTs) increasingly adopt large expansion cohorts exploring novel agents across different tumor types. High-grade glioma (HGG) patients are usually excluded from these trials. METHODS: Data of patients with recurrent HGGs treated within multicohort ieCTs between February 2014 and August 2019 (experimental group, EG) at our Phase I Unit were retrospectively reviewed and compared to a matched control group (CG) of patients treated with standard therapies. We retrospectively evaluated clinical, laboratory, and molecular parameters through univariate and multivariate analysis. A prospective characterization of circulating leukocyte subpopulations was performed in the latest twenty patients enrolled in the EG, with a statistical significance cutoff of P < .1. RESULTS: Thirty HGG patients were treated into six ieCTs. Fifteen patients received monotherapies (anti-PD-1, anti-CSF-1R, anti-TGFß, anti-cereblon), fifteen patients combination regimens (anti-PD-L1 + anti-CD38, anti-PD-1 + anti-CSF-1R). In the EG, median progression-free survival and overall survival (OS) from treatment initiation were 1.8 and 8.6 months; twelve patients survived more than 12 months, and two of them more than 6 years. Univariate analysis identified O 6-methylguanine DNA methyltransferase (MGMT) promoter methylation and total protein value at six weeks as significantly correlated with a better outcome. Decreased circulating neutrophils and increased conventional dendritic cells levels lead to significantly better OS. CONCLUSIONS: A subgroup of EG patients achieved remarkably durable disease control. MGMT promoter methylation identifies patients who benefit more from immunotherapy. Monitoring dynamic changes of innate immune cell populations may help to predict clinical outcomes.

5.
Cell Rep ; 37(3): 109871, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686325

RESUMEN

Human Vδ2 cells are innate-like γδ T effectors performing potent immune surveillance against tumors. The constitutive expression of NKG2A identifies a subset of Vδ2 T cells licensed with an intrinsic hyper-responsiveness against cancer. Indeed, the transcriptomic profiles of NKG2A+ and NKG2A- cells characterize two distinct "intralineages" of Vδ2 T lymphocytes that appear early during development, keep their phenotypes, and show self-renewal capabilities in adult life. The hyper-responsiveness of NKG2A+ Vδ2 T cells is counterbalanced by the inhibitory signaling delivered by human leukocyte antigen E (HLA-E) expressed on malignant cells as a tumor-escape mechanism. However, either masking or knocking out NKG2A restores the capacity of Vδ2 T cells to exert the highest effector functions even against HLA-E+ tumors. This is highly relevant in the clinic, as the different degrees of engagement of the NKG2A-HLA-E checkpoint in hepatocellular carcinoma, glioblastoma, and non-small cell lung cancer directly impact patients' overall survival. These findings open avenues for developing combined cellular and immunologic anticancer therapies.


Asunto(s)
Citotoxicidad Inmunológica , Linfocitos Intraepiteliales/metabolismo , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Anciano , Estudios de Casos y Controles , Proliferación Celular , Autorrenovación de las Células , Técnicas de Cocultivo , Citocinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata , Lactante , Linfocitos Intraepiteliales/inmunología , Células K562 , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Subfamília C de Receptores Similares a Lectina de Células NK/genética , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Fenotipo , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal
6.
Cancers (Basel) ; 13(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062821

RESUMEN

Natural killer (NK) and dendritic cells (DCs) are innate immune cells that play a crucial role in anti-tumor immunity. NK cells kill tumor cells through direct cytotoxicity and cytokine secretion. DCs are needed for the activation of adaptive immune responses against tumor cells. Both NK cells and DCs are subdivided in several subsets endowed with specialized effector functions. Crosstalk between NK cells and DCs leads to the reciprocal control of their activation and polarization of immune responses. In this review, we describe the role of NK cells and DCs in liver cancer, focusing on the mechanisms involved in their reciprocal control and activation. In this context, intrahepatic NK cells and DCs present unique immunological features, due to the constant exposure to non-self-circulating antigens. These interactions might play a fundamental role in the pathology of primary liver cancer, namely hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Additionally, the implications of these immune changes are relevant from the perspective of improving the cancer immunotherapy strategies in HCC and ICC patients.

7.
Ital J Dermatol Venerol ; 156(3): 356-365, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33179877

RESUMEN

Kaposi's sarcoma (KS) is a lymphangioproliferative disorder associated with Human herpesvirus 8 (HHV8) infection. Four clinical subtypes are recognized: classic, endemic, epidemic (HIV-related) and iatrogenic. KS diagnosis is based on clinical features, histopathological assessment, and HHV8 serology. Classic KS is usually skin-limited and has a chronic course, while the iatrogenic variant may show mucosal, nodal or visceral involvement. Clinical staging is fundamental to guide the management. Localized disease may be treated with different local therapies, even if there are no randomized trials comparing these different modalities. Aggressive, disseminated KS and cases with visceral involvement usually require systemic chemotherapy, most commonly vinblastine, bleomycin or paclitaxel. Iatrogenic KS needs immunosuppression tapering/withdrawal and, if possible, switch to m-TOR inhibitors in post-transplant KS. The present work by a panel of Italian experts provides guidelines on KS diagnosis and management based on a critical review of the literature and a long and extensive personal experience.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida , Sarcoma de Kaposi , Neoplasias Cutáneas , Humanos , Enfermedad Iatrogénica , Italia/epidemiología , Sarcoma de Kaposi/diagnóstico , Neoplasias Cutáneas/diagnóstico
8.
Cytometry A ; 99(3): 218-230, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33098618

RESUMEN

Dendritic cells (DCs) play a crucial role in the complex interplay between tumor cells and the immune system. During the elimination phase of cancer immunoediting, immunostimulatory DCs are critical for the control of tumor growth. During the escape phase, regulatory DCs sustain tumor tolerance and contribute to the development of the immunosuppressive tumor microenvironment that characterizes this phase. Moreover, increasing evidence indicates that DCs are also critical for the success of cancer immunotherapy. Hence, there is increasing need to fully characterize DC subsets and their activatory/inhibitory profile in cancer patients. In this review, we describe the role played by different DC subsets in the different phases of cancer immunoediting, the function exerted by different activatory and inhibitory molecules expressed on DC surface, and the cytokines produced by distinct DC subsets, in order to provide an overview on the DC features that may be useful to be assessed when dealing with the flow cytometric characterization of DCs in cancer patients. © 2020 International Society for Advancement of Cytometry.


Asunto(s)
Células Dendríticas , Neoplasias , Citometría de Flujo , Humanos , Tolerancia Inmunológica , Inmunoterapia , Microambiente Tumoral
9.
Immunobiology ; 225(1): 151853, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31703822

RESUMEN

Glioma is the most common primary brain cancer, and half of patients present a diagnosis of glioblastoma (GBM), its most aggressive and lethal form. Conventional therapies, including surgery, radiotherapy, and chemotherapy, have not resulted in major ameliorations in GBM survival outcome, which remains extremely poor. Recent immunotherapy improvements for other tumors, coupled with growing knowledge of the complex interactions between malignant glioma cells and the immune system, led to an exponential increase in glioma immunotherapy research. However, immunotherapeutic strategies in GBM have not yet reached their full potential, mainly due to the limited understanding of the strong immunosuppressive microenvironment (TME) characterizing this tumor. Glioma-associated macrophages and microglia (GAMs) are key drivers of the local immunosuppression promoting tumor progression and its resistance to immunomodulating therapeutic strategies. Together with other myeloid cells, such as dendritic cells and neutrophils, GAMs actively shape glioma TME, modulate anti-tumoral immune response and support angiogenesis, tumor cell invasion and proliferation. In this review, we discuss the role of myeloid cells in the complex TME of glioma and the available clinical data on therapeutic strategies focusing on approaches that affect myeloid cells activity in GBM.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioma/inmunología , Macrófagos/inmunología , Microglía/inmunología , Células Supresoras de Origen Mieloide/fisiología , Animales , Terapia Biológica , Humanos , Terapia de Inmunosupresión , Microambiente Tumoral
10.
Cardiovasc Res ; 116(3): 698-707, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31135876

RESUMEN

AIMS: The pathogenetic mechanisms underlying unprovoked venous thromboembolism (uVTE) are largely unknown. In this study, we investigated the molecular mechanisms involved in uVTE pathogenesis by using ex vivo expanded endothelial colony-forming cells (ECFCs), which represent a valuable non-invasive tool for the assessment of endothelial function. METHODS AND RESULTS: We isolated and expanded ECFCs from the peripheral blood of uVTE patients and observed that these cells underwent earlier senescence and showed lower growth rate compared with ECFCs obtained from healthy donors. Through microarray expression profiling, we demonstrated that 2905 genes were differentially expressed between patients and controls. Among them, the anti-angiogenic cytokine TNF superfamily member 15 (TNFSF15) and its death-receptor TNFRSF25 were up-regulated in uVTE ECFCs, and this finding was validated by RT-qPCR. TNFSF15 up-regulation was confirmed at the protein level in ECFC supernatants, and the in vivo relevance of these findings was further corroborated by demonstrating that also the plasmatic levels of TNFSF15 are increased in uVTE patients. After proving that exogenous TNFSF15 exerts pro-apoptotic and anti-proliferative activity on control ECFCs, we demonstrated through blocking experiments that TNFSF15 up-regulation contributes to impaired survival and proliferation of uVTE ECFCs. CONCLUSION: By providing evidence that TNFSF15 impairs ECFC functions crucial to endothelial repair, and that uVTE patients have increased TNFSF15 levels both ex vivo and in vivo, the results of this study suggest that pathologic up-regulation of TNFSF15-TNFRSF25 axis may contribute to uVTE pathogenesis, and may represent the target for novel therapeutic strategies aimed at preventing recurrences in uVTE patients.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Tromboembolia Venosa/metabolismo , Adulto , Apoptosis , Estudios de Casos y Controles , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Senescencia Celular , Células Progenitoras Endoteliales/patología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Transducción de Señal , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Tromboembolia Venosa/patología , Tromboembolia Venosa/fisiopatología
11.
JCI Insight ; 4(24)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31689241

RESUMEN

γδ T cells account for a large fraction of human intestinal intraepithelial lymphocytes (IELs) endowed with potent antitumor activities. However, little is known about their origin, phenotype, and clinical relevance in colorectal cancer (CRC). To determine γδ IEL gut specificity, homing, and functions, γδ T cells were purified from human healthy blood, lymph nodes, liver, skin, and intestine, either disease-free, affected by CRC, or generated from thymic precursors. The constitutive expression of NKp46 specifically identifies a subset of cytotoxic Vδ1 T cells representing the largest fraction of gut-resident IELs. The ontogeny and gut-tropism of NKp46+/Vδ1 IELs depends both on distinctive features of Vδ1 thymic precursors and gut-environmental factors. Either the constitutive presence of NKp46 on tissue-resident Vδ1 intestinal IELs or its induced expression on IL-2/IL-15-activated Vδ1 thymocytes are associated with antitumor functions. Higher frequencies of NKp46+/Vδ1 IELs in tumor-free specimens from CRC patients correlate with a lower risk of developing metastatic III/IV disease stages. Additionally, our in vitro settings reproducing CRC tumor microenvironment inhibited the expansion of NKp46+/Vδ1 cells from activated thymic precursors. These results parallel the very low frequencies of NKp46+/Vδ1 IELs able to infiltrate CRC, thus providing insights to either follow-up cancer progression or to develop adoptive cellular therapies.


Asunto(s)
Neoplasias Colorrectales/inmunología , Mucosa Intestinal/citología , Linfocitos Intraepiteliales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Linfocitos T Citotóxicos/inmunología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Antígenos Ly/metabolismo , Colon/citología , Colon/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Progresión de la Enfermedad , Femenino , Humanos , Íleon/citología , Íleon/inmunología , Inmunoterapia Adoptiva/métodos , Mucosa Intestinal/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/trasplante , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Globulina de Unión a Hormona Sexual , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/trasplante , Adulto Joven
12.
Front Immunol ; 10: 1325, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31244860

RESUMEN

Dendritic cells (DCs) play a crucial role in initiating and shaping immune responses. The effects of DCs on adaptive immune responses depend partly on functional specialization of distinct DC subsets, and partly on the activation state of DCs, which is largely dictated by environmental signals. Fully activated immunostimulatory DCs express high levels of costimulatory molecules, produce pro-inflammatory cytokines, and stimulate T cell proliferation, whereas tolerogenic DCs express low levels of costimulatory molecules, produce immunomodulatory cytokines and impair T cell proliferation. Relevant to the increasing use of immune checkpoint blockade in cancer treatment, signals generated from inhibitory checkpoint molecules on DC surface may also contribute to the inhibitory properties of tolerogenic DCs. Yet, our knowledge on the expression of inhibitory molecules on human DC subsets is fragmentary. Therefore, in this study, we investigated the expression of three immune checkpoints on peripheral blood DC subsets, in basal conditions and upon exposure to pro-inflammatory and anti-inflammatory stimuli, by using a flow cytometric panel that allows a direct comparison of the activatory/inhibitory phenotype of DC-lineage and inflammatory DC subsets. We demonstrated that functionally distinct DC subsets are characterized by differential expression of activatory and inhibitory molecules, and that cDC1s in particular are endowed with a unique immune checkpoint repertoire characterized by high TIM-3 expression, scarce PD-L1 expression and lack of ILT2. Notably, this unique cDC1 repertoire was subverted in a group of patients with myelodysplastic syndromes included in the study. Applied to the characterization of DCs in the tumor microenvironment, this panel has the potential to provide valuable information to be used for investigating the role of DC subsets in cancer, guiding DC-targeting treatments, and possibly identifying predictive biomarkers for clinical response to cancer immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Adaptativa , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Antígeno B7-H1/metabolismo , Linaje de la Célula/inmunología , Citocinas/biosíntesis , Células Dendríticas/clasificación , Células Dendríticas/metabolismo , Femenino , Citometría de Flujo/métodos , Voluntarios Sanos , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/inmunología , Adulto Joven
13.
Eur J Dermatol ; 28(3): 320-325, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30105990

RESUMEN

The relationship between Kaposi's sarcoma (KS) and psoriasis is still controversial. To analyse the association between KS and psoriasis, address the hypothesis of a reciprocal influence between the two conditions relative to clinical presentation and evolution, and consider the best therapeutic approach to be used for the treatment of psoriasis in KS patients in order to avoid the typical induction or worsening of KS during immunosuppression. We retrospectively reviewed clinical records of 37 patients with KS and psoriasis. Fisher's exact test was performed in order to compare epidemiological and clinical data between subsets of patients. The prevalence of psoriasis in our KS population (n = 1407) was 2.6%. There were no statistically significant differences in terms of stage or rate of progression between KS patients with and without psoriasis, except for a higher frequency of patients with KS Stage IIB among patients with KS and psoriasis (p = 0.001). Patients with psoriasis have a risk of KS comparable to that of the general population. Psoriasis and KS do not appear to influence each other. For the treatment of psoriasis in KS patients, one should take into account the KS-inducing potential of certain anti-psoriatic drugs.


Asunto(s)
Psoriasis/complicaciones , Sarcoma de Kaposi/complicaciones , Adolescente , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Humanos , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Psoriasis/tratamiento farmacológico , Psoriasis/inmunología , Estudios Retrospectivos , Factores de Riesgo , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/patología , Adulto Joven
14.
FASEB J ; 32(7): 3742-3751, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29436863

RESUMEN

Diabetic neuropathy is the most common complication of diabetes and is frequently associated with foot ischemia and infection, but its pathogenesis is controversial. We hypothesized that proinsulin expression in peripheral blood mononuclear cells is a process relevant to this condition and could represent a link among hyperglycemia, nerve susceptibility, and diabetic foot lesions. We assessed proinsulin expression by using flow cytometry in dendritic cells from control participants and patients with type 2 diabetes with or without peripheral neuropathy or accompanied by diabetic foot. Among 32 non-neuropathic and 120 neuropathic patients with type 2 diabetes, we performed leg electromyography and found average sensory sural nerve conduction velocities of 48 ± 4 and 30 ± 4 m/s, respectively ( P < 0.03). Of those with neuropathy, 42 were without lesions, 39 had foot lesions, and 39 had neuroischemic foot lesions (allux oximetry <30 mmHg). In this well-defined diabetic population, but not in nondiabetic participants, a progressively increasing level of peripheral blood dendritic cell proinsulin expression was detected, which directly correlated with circulating TNF-α levels ( P < 0.002) and multiple conduction velocities of leg nerves ( P < 0.05). These results are consistent with the hypothesis that, in type 2 diabetes, proinsulin-expressing blood cells, possibly via their involvement in innate immunity, may play a role in diabetic peripheral neuropathy and foot lesions.-Sambataro, M., Sambado, L., Trevisiol, E., Cacciatore, M., Furlan, A., Stefani, P. M., Seganfreddo, E., Durante, E., Conte, S., Della Bella, S., Paccagnella, A., dei Tos, A. P. Proinsulin-expressing dendritic cells in type 2 neuropathic diabetic patients with and without foot lesions.


Asunto(s)
Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Pie Diabético/metabolismo , Neuropatías Diabéticas/metabolismo , Proinsulina/metabolismo , Pie Diabético/patología , Pie Diabético/fisiopatología , Neuropatías Diabéticas/patología , Neuropatías Diabéticas/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nervio Sural/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Leukoc Biol ; 103(3): 525-533, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29345349

RESUMEN

Endothelial colony-forming cells (ECFCs), a unique endothelial stem cell population, are highly increased in the blood of Kaposi sarcoma (KS) patients. KS-derived ECFCs (KS-ECFCs) are also endowed with increased proliferative and vasculogenic potential, thus suggesting that they may be precursors of KS spindle cells. However, the mechanisms underlying the increased proliferative activity of KS-ECFCs remain poorly understood. Sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) are metabolically interconnected sphingoid mediators crucial to cell proliferation. Here, we investigated the metabolism, release, and proliferative effects of S1P and C1P in KS-ECFCs compared with control ECFCs (Ct-ECFCs). Metabolic studies by cell labeling, chromatographic analyses, and digital autoradiography revealed that S1P and C1P biosynthesis and S1P secretion are all efficient processes in KS-ECFCs, more efficient in KS-ECFCs than Ct-ECFCs. Quantitative PCR analyses demonstrated a significantly higher ceramide kinase and sphingosine kinase-2 expression in KS-ECFCs. Notably, also the expression of S1P1 and S1P3 receptors was augmented in KS-ECFCs. Accordingly, treatment with exogenous C1P or S1P induced a significant, concentration-dependent stimulation of KS-ECFC proliferation, but was almost completely ineffective in Ct-ECFCs. Hence, we identified C1P and S1P as autocrine/paracrine proliferative signals in KS-ECFCs. A better understanding of the mechanisms that enhance S1P/C1P formation in KS-ECFCs may yield effective therapeutic modalities.


Asunto(s)
Proliferación Celular , Ceramidas/metabolismo , Endotelio Vascular/patología , Lisofosfolípidos/metabolismo , Sarcoma de Kaposi/patología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Diferenciación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas de Unión al ARN/metabolismo , Sarcoma de Kaposi/metabolismo , Transducción de Señal
17.
Gastroenterology ; 153(5): 1363-1377.e6, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28827082

RESUMEN

BACKGROUND & AIMS: Alterations in signaling pathways that regulate resolution of inflammation (resolving pathways) contribute to pathogenesis of ulcerative colitis (UC). The resolution process is regulated by lipid mediators, such as those derived from the ω-3 docosahexaenoic acid (DHA), whose esterified form is transported by the major facilitator superfamily domain containing 2A (MFSD2A) through the endothelium of brain, retina, and placenta. We investigated if and how MFSD2A regulates lipid metabolism of gut endothelial cells to promote resolution of intestinal inflammation. METHODS: We performed lipidomic and functional analyses of MFSD2A in mucosal biopsies and primary human intestinal microvascular endothelial cells (HIMECs) isolated from surgical specimens from patients with active, resolving UC and healthy individuals without UC (controls). MFSD2A was knocked down in HIMECs with small hairpin RNAs or overexpressed from a lentiviral vector. Human circulating endothelial progenitor cells that overexpress MFSD2A were transferred to CD1 nude mice with dextran sodium sulfate-induced colitis, with or without oral administration of DHA. RESULTS: Colonic biopsies from patients with UC had reduced levels of inflammation-resolving DHA-derived epoxy metabolites compared to healthy colon tissues or tissues with resolution of inflammation. Production of these metabolites by HIMECs required MFSD2A, which is required for DHA retention and metabolism in the gut vasculature. In mice with colitis, transplanted endothelial progenitor cells that overexpressed MFSD2A not only localized to the inflamed mucosa but also restored the ability of the endothelium to resolve intestinal inflammation, compared with mice with colitis that did not receive MFSD2A-overexpressing endothelial progenitors. CONCLUSIONS: Levels of DHA-derived epoxides are lower in colon tissues from patients with UC than healthy and resolving mucosa. Production of these metabolites by gut endothelium requires MFSD2A; endothelial progenitor cells that overexpress MFSD2A reduce colitis in mice. This pathway might be induced to resolve intestinal inflammation in patients with colitis.


Asunto(s)
Colitis/prevención & control , Colon/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Células Progenitoras Endoteliales/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colon/efectos de los fármacos , Colon/patología , Sistema Enzimático del Citocromo P-450/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Compuestos Epoxi/metabolismo , Humanos , Proteínas de Transporte de Membrana/genética , Ratones Desnudos , Oxilipinas/metabolismo , Interferencia de ARN , Transducción de Señal , Simportadores , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Supresoras de Tumor/genética
18.
J Invest Dermatol ; 137(7): 1533-1540, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28288847

RESUMEN

Kaposi's sarcoma (KS) is characterized by hyperproliferation of spindle cells that have an endothelial origin and assume their characteristic features upon infection with human herpesvirus-8, the causative agent for KS. The multifocal nature of KS suggests that spindle cells derive from circulating HHV8-infected precursors that yet lack identification. We investigated whether endothelial colony-forming cells (ECFCs) obtained from KS patients may be putative precursors of spindle cells by assessing whether their in vitro behavior may evoke the in vivo behavior of KS spindle cells. We isolated and cultured ECFCs from the blood of 83 patients with classic KS and compared them with ECFCs obtained from 86 healthy donors. ECFCs were highly increased in the blood of classic KS patients; they showed higher proliferative and vasculogenic potential and higher production of IL-6 than control ECFCs. Similarly to spindle cells in KS lesions, a variable proportion of cells within each ECFC colony expressed the human herpesvirus-8 latency-associated nuclear antigen. ECFCs obtained from classic KS patients evoked KS spindle cell behavior, thus supporting the hypothesis that ECFCs may be putative precursors of spindle cells. ECFCs can therefore represent a noninvasive tool for studying KS and screening drug activity, thus possibly guiding personalized care for KS patients.


Asunto(s)
Células Endoteliales/patología , Interleucina-6/metabolismo , Sarcoma de Kaposi/patología , Neoplasias Cutáneas/patología , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sarcoma de Kaposi/metabolismo , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas , Adulto Joven
19.
J Leukoc Biol ; 99(1): 67-78, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26538528

RESUMEN

Preterm birth is the leading cause of neonatal morbidity and mortality. Although the underlying causes of pregnancy-associated complication are numerous, it is well established that infection and inflammation represent a highly significant risk factor in preterm birth. However, despite the clinical and public health significance, infectious agents, molecular trigger(s), and immune pathways underlying the pathogenesis of preterm birth remain underdefined and represent a major gap in knowledge. Here, we provide an overview of recent clinical and animal model data focused on the interplay between infection-driven inflammation and induction of preterm birth. Furthermore, here, we highlight the critical gaps in knowledge that warrant future investigations into the interplay between immune responses and induction of preterm birth.


Asunto(s)
Inflamación/complicaciones , Nacimiento Prematuro/etiología , Animales , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata , Infecciones/complicaciones , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Embarazo , Nacimiento Prematuro/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Toll-Like/metabolismo
20.
Rheumatology (Oxford) ; 54(8): 1429-34, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25740829

RESUMEN

OBJECTIVE: The aim of this study was to investigate the biological effects of belimumab on B cells in the first phase II open-label trial with belimumab in patients with primary SS (pSS) (BELISS). METHODS: Peripheral blood B cell subsets and their B cell activating factor-receptor (BAFF-R) expression were analysed by multicolour flow cytometry in 10 pSS patients either before or after 24 and 52 weeks of therapy with belimumab. Serum BAFF levels were analysed by ELISA. RESULTS: At baseline, pSS patients showed a significant increase in circulating B cells compared with healthy donors matched for age and sex, with a predominant expansion of transitional and naive B cell subsets. pSS patients also showed higher serum BAFF levels and lower B cell BAFF-R expression. Therapy with belimumab in pSS patients induced a significant reduction in transitional and naive B cell subsets to levels similar to those observed in healthy donors. Furthermore, belimumab normalized BAFF-R expression in all B subsets comprised within the memory compartment. The restoration of B cell frequency and subset composition in response to belimumab was also associated with a decrease in serum levels of Ig, RF, ANAs, and with an increase in the C4 complement fraction. All of these belimumab-mediated effects were observed after 24 weeks of therapy and maintained until the end of the therapeutic protocol. CONCLUSION: Taken together, our findings show that targeting BAFF with belimumab is successful in normalizing B cell frequency, phenotype and functions in pSS. TRIAL REGISTRATION: clinicaltrials.gov; https://clinicaltrials.gov/; NCT01008982.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Receptor del Factor Activador de Células B/metabolismo , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/metabolismo , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/metabolismo , Adulto , Anciano , Factor Activador de Células B/metabolismo , Subgrupos de Linfocitos B/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Femenino , Homeostasis , Humanos , Inmunoglobulina G/sangre , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Persona de Mediana Edad , Factor Reumatoide/sangre , Síndrome de Sjögren/patología , Resultado del Tratamiento , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA